Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 7_Supplement ( 2023-04-04), p. 5037-5037
    Abstract: The development of effective radioligand therapeutics (RLTs) is frequently hampered by the lack of high-quality targeting agents that selectively deliver radioactive payloads to the site of disease while sparing healthy tissues. Antibodies can have high affinity and specificity to tumor targets, but their large size results in limited tumor penetration and long systemic half-life is frequently causing haematological toxicities. Alternatively, targeting agents with low molecular weight such as small molecules and peptides often suffer from limited affinity and specificity to the tumor target, resulting in off-target effects and limited tumor retention. DARPins (Designed Ankyrin Repeat Proteins) developed by Molecular Partners combine small size (15 kDa) and ideal binding properties. Due to their rigid-body target binding mode DARPins combine very high affinity and specificity and unless engineered accordingly, DARPins have very short systemic half-lives. Thanks to a simple and robust architecture, DARPins can be efficiently coupled with radioactive payloads, even at elevated temperatures; and they can tolerate sequence-engineering approaches, which are not compatible with other protein scaffolds. To establish the DARPin platform for RLT, we have used DARPin candidates against different tumor targets. We have previously shown that increasing affinity to the tumor target correlates with elevated tumor uptake and long tumor residence in preclinical mouse models. We now also show that DARPins exhibit a homogeneous and deep tumor penetration in vivo that is highly superior to antibody benchmarks. Globular proteins below 60 kDa in size are typically cleared from the bloodstream via the renal pathway. This generally results in a strong kidney accumulation of small sized, protein-based targeting agents and their coupled residualizing radionuclides, leading to dose-limiting kidney toxicities. To overcome this limitation, we have undertaken an extensive engineering approach of the DARPin scaffold. Our results show that sequence engineering strongly reduces kidney uptake of DARPins without affecting their tumor uptake. This effect was confirmed with independent DARPin candidates suggesting a general applicability of the approach. Combined with other orthogonal strategies, we are able obtain favourable tumor to kidney ratios in preclinical mouse models. These results show that our proprietary optimized DARPin platform offers an attractive solution to the limitations of protein-based targeting agents for RLT applications. Together with the fact that high-affinity DARPins can be generated against a large variety of tumor targets, we conclude that our platform provides a powerful basis for the development of next-generation RLTs. Several DARPin-RLT programs in indications with high unmet medical need are currently in development. Citation Format: Andreas Bosshart, Stephan Wullschleger, Martin Behe, Alain Blanc, Stefan Imobersteg, Alexandra Neculcea, Jacqueline Blunschi, Liridon Abduli, Sarah Schütz, Julia Wolter, Christian Reichen, Amelie Croset, Alessandra Villa, Christian Lizak, Anne Goubier, Roger Schibli, Daniel Steiner. DARPins as powerful targeting agents for radioligand therapeutics. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; C ancer Res 2023;83(7_Suppl):Abstract nr 5037.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Nature Cell Biology, Springer Science and Business Media LLC, Vol. 14, No. 8 ( 2012-8), p. 882-890
    Type of Medium: Online Resource
    ISSN: 1465-7392 , 1476-4679
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2012
    detail.hit.zdb_id: 1494945-3
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 2891-2891
    Abstract: Intracellular tumor antigens presented as peptides on MHC (pMHC) class I molecules are attractive targets for more tumor-selective immunotherapeutic approaches with promising data already emerging from clinical trials. pMHCs have been targeted by TCR-engineered T cells or soluble recombinant T-cell receptors (TCRs) fused to an anti-CD3 fragment. Naturally occurring cancer-reactive TCRs have weak affinity and require substantial affinity enhancements for their cognate pMHC. However, the outcome of this process is difficult to predict and bears the risk for off-target cross reactivities in normal tissues, which may lead to severe adverse events in the clinic. Here, we describe a highly potent dual pMHC T-cell engager (TCE) antibody format with high specificity towards tumor-specific pMHCs utilizing an HLA-A*02:01 restricted MAGE-A4 epitope to validate the concept. A series of monovalent and bivalent antibody constructs composed of anti-MAGE-A4 binding arms, ranging in affinities from 30 nM to 100 pM, were fused to an anti-CD3 Fab fragment with lower affinity compared to that commonly used for TCR-fusions. The different antibody constructs were evaluated for selective killing of MAGE-A4/HLA-A*02 positive human U2OS osteosarcoma and A375 melanoma cancer cells versus a panel of different MAGE-A4-negative/HLA-A*02-positive human cell lines. Bivalent bispecific antibody variants mediated a 7-fold greater degree of cancer cell killing and similarly increased T cell activation compared to their monovalent bispecific counterparts. EC50 values ranged as low as single digit picomolar, while the overall cross reactivity against MAGE-A4-negative/HLA-A*02-positive cells was not substantially affected. These results prove that dual targeting of pMHCs on cancer cells provides selective and efficient T cell-mediated target cell killing and T cell activation, even at very low levels of pMHC on the cell surface, highlighting the pivotal roles played by the affinity of the individual arms, valency, and epitope densities. We further analyzed the dual pMHC TCE for potential off-target effects by recognition of similar and physiologically relevant non-MAGE-A4 peptides. T2 cells pulsed with similar peptides and co-cultured with PBMC effector cells showed no significant T cell activation or IFNg release in the presence of the dual pMHC TCE in comparison to MAGE-A4 peptide-pulsed T2 cells. Finally, we compared the dual pMHC TCE against a recombinant TCR fused to an anti-CD3 scFv, a construct that is currently in clinical development. Interestingly, the dual pMHC TCE resulted in a 3-fold more potent cancer cell killing while having significantly lower effect on cytokine production. In conclusion, dual pMHC targeting with TCE antibodies is an attractive alternative to soluble affinity-enhanced TCR-based cancer immunotherapies as they facilitate potent tumor targeting without the need for extensive affinity enhancements. Citation Format: Stephanie Jungmichel, Fabian Scheifele, Anna Sobieraj, Severin Wendelspiess, Thomas Schleier, Philip Knobel, Camilla Winnewisser, Melissa Vrohlings, Philipp Richle, Hannes Merten, Jacqueline Blunschi, Christian Leisner, Leonardo Borras. Enhanced anti-tumor responses with a novel dual pMHC T-cell engager bispecific antibody [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 2891.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages